Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Faraday Discuss ; 187: 43-68, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27095431

ABSTRACT

Breast cancer screening provides sensitive tumor identification, but low specificity implies that a vast majority of biopsies are not ultimately diagnosed as cancer. Automated techniques to evaluate biopsies can prevent errors, reduce pathologist workload and provide objective analysis. Fourier transform infrared (FT-IR) spectroscopic imaging provides both molecular signatures and spatial information that may be applicable for pathology. Here, we utilize both the spectral and spatial information to develop a combined classifier that provides rapid tissue assessment. First, we evaluated the potential of IR imaging to provide a diagnosis using spectral data alone. While highly accurate histologic [epithelium, stroma] recognition could be achieved, the same was not possible for disease [cancer, no-cancer] due to the diversity of spectral signals. Hence, we employed spatial data, developing and evaluating increasingly complex models, to detect cancers. Sub-mm tumors could be very confidently predicted as indicated by the quantitative measurement of accuracy via receiver operating characteristic (ROC) curve analyses. The developed protocol was validated with a small set and statistical performance used to develop a model that predicts study design for a large scale, definitive validation. The results of evaluation on different instruments, at higher noise levels, under a coarser spectral resolution and two sampling modes [transmission and transflection], indicate that the protocol is highly accurate under a variety of conditions. The study paves the way to validating IR imaging for rapid breast tumor detection, its statistical validation and potential directions for optimization of the speed and sampling for clinical deployment.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Spectroscopy, Fourier Transform Infrared/methods , Humans , Reproducibility of Results , Sensitivity and Specificity
2.
Breast Cancer Res Treat ; 145(3): 593-604, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24810497

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that lacks effective targeted therapies. The epithelial-to-mesenchymal transition (EMT) is a key contributor in the metastatic process. We previously showed the pan-deacetylase inhibitor LBH589 induces CDH1 expression in TNBC cells, suggesting regulation of EMT. The purpose of this study was to examine the effects of LBH589 on the metastatic qualities of TNBC cells and the role of EMT in this process. A panel of breast cancer cell lines (MCF-7, MDA-MB-231, and BT-549), drugged with LBH589, was examined for changes in cell morphology, migration, and invasion in vitro. The effect on in vivo metastasis was examined using immunofluorescent staining of lung sections. EMT gene expression profiling was used to determine LBH589-induced changes in TNBC cells. ZEB overexpression studies were conducted to validate requirement of ZEB in LBH589-mediated proliferation and tumorigenesis. Our results indicate a reversal of EMT by LBH589 as demonstrated by altered morphology and altered gene expression in TNBC. LBH589 was shown to be a more potent inhibitor of EMT than other HDAC inhibitors, SAHA and TMP269. Additionally, we found that LBH589 inhibits metastasis of MDA-MB-231 cells in vivo. These effects of LBH589 were mediated in part by inhibition of ZEB, as overexpression of ZEB1 or ZEB2 mitigated the effects of LBH589 on MDA-MB-231 EMT-associated gene expression, migration, invasion, CDH1 expression, and tumorigenesis. These data indicate therapeutic potential of LBH589 in targeting EMT and metastasis of TNBC.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Homeodomain Proteins/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Repressor Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Homeodomain Proteins/biosynthesis , Humans , MCF-7 Cells , Mice , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/drug therapy , Panobinostat , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Xenograft Model Antitumor Assays , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
3.
Breast Cancer Res ; 14(3): R79, 2012 May 21.
Article in English | MEDLINE | ID: mdl-22613095

ABSTRACT

INTRODUCTION: Of the more than one million global cases of breast cancer diagnosed each year, approximately fifteen percent are characterized as triple-negative, lacking the estrogen, progesterone, and Her2/neu receptors. Lack of effective therapies, younger age at onset, and early metastatic spread have contributed to the poor prognoses and outcomes associated with these malignancies. Here, we investigate the ability of the histone deacetylase inhibitor panobinostat (LBH589) to selectively target triple-negative breast cancer (TNBC) cell proliferation and survival in vitro and tumorigenesis in vivo. METHODS: TNBC cell lines MDA-MB-157, MDA-MB-231, MDA-MB-468, and BT-549 were treated with nanomolar (nM) quantities of panobinostat. Relevant histone acetylation was verified by flow cytometry and immunofluorescent imaging. Assays for trypan blue viability, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) proliferation, and DNA fragmentation were used to evaluate overall cellular toxicity. Changes in cell cycle progression were assessed with propidium iodide flow cytometry. Additionally, qPCR arrays were used to probe MDA-MB-231 cells for panobinostat-induced changes in cancer biomarkers and signaling pathways. Orthotopic MDA-MB-231 and BT-549 mouse xenograft models were used to assess the effects of panobinostat on tumorigenesis. Lastly, flow cytometry, ELISA, and immunohistochemical staining were applied to detect changes in cadherin-1, E-cadherin (CDH1) protein expression and the results paired with confocal microscopy in order to examine changes in cell morphology. RESULTS: Panobinostat treatment increased histone acetylation, decreased cell proliferation and survival, and blocked cell cycle progression at G2/M with a concurrent decrease in S phase in all TNBC cell lines. Treatment also resulted in apoptosis induction at 24 hours in all lines except the MDA-MB-468 cell line. MDA-MB-231 and BT-549 tumor formation was significantly inhibited by panobinostat (10 mg/kg/day) in mice. Additionally, panobinostat up-regulated CDH1 protein in vitro and in vivo and induced cell morphology changes in MDA-MB-231 cells consistent with reversal of the mesenchymal phenotype. CONCLUSIONS: This study revealed that panobinostat is overtly toxic to TNBC cells in vitro and decreases tumorigenesis in vivo. Additionally, treatment up-regulated anti-proliferative, tumor suppressor, and epithelial marker genes in MDA-MB-231 cells and initiated a partial reversal of the epithelial-to-mesenchymal transition. Our results demonstrate a potential therapeutic role of panobinostat in targeting aggressive triple-negative breast cancer cell types.


Subject(s)
Breast Neoplasms/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cdh1 Proteins , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression , Histones/metabolism , Humans , Mice , Mice, SCID , Panobinostat , Random Allocation , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...